As is well established in invasive breast disease, it is becoming increasingly clear that molecular heterogeneity, both between and within lesions, is a prevalent, distinct phenotype of premalignant lesions of the breast. Key pathways of tumorigenesis modulate critical features of premalignant lesions such as proliferation, differentiation, stress response, and even the generation of diversity. Current studies show that evaluation of these lesions may provide clinically useful information on future tumor formation as well as biological insights into the origin and functional significance of this distinct phenotype. Cancer Prev Res; 3(5); 579–87. ©2010 AACR.

Premalignant lesions of the breast are becoming an increasing focus of clinical breast care and translational and basic science. Detection of these lesions has increased because of widespread screening programs and increasing numbers of biopsies. Although a premalignancy diagnosis is a risk factor for future development of invasive breast cancer, the majority of so-diagnosed individuals will remain free of invasive disease following excision of the lesion. Therefore, continued scientific insights into the origins and behavior of in situ carcinomas and atypical lesions are essential to achieving the goal of distinguishing the higher-risk minority of patients, who are destined to develop invasive carcinomas and thus need aggressive prevention and screening, from the lower-risk majority of patients, who could be directed to more limited interventions.

Particularly promising efforts toward realizing this goal of risk stratification are recent studies to elucidate the molecular heterogeneity of premalignant lesions such as ductal carcinoma in situ (DCIS). DCIS has long been known to be heterogeneous in its morphologic features, presentation, and clinical behavior. The emergence of genomic and gene expression profiling has established that DCIS is heterogeneous at a level of molecular complexity that is comparable with or perhaps greater than that of invasive disease (110). Although certainly less studied, the heterogeneity observed in the microenvironment surrounding individual premalignant lesions may also be a critical determinant of their future behavior. Recent studies have begun to identify heterogeneity within the microenvironment surrounding invasive tumors that could be linked to outcome (1114). The genesis of these stromal changes may precede the formation of invasive lesions and reflect causes or consequences of early tumor progression. This discussion will focus on the epithelial cells within a premalignant lesion and the biological and clinical ramifications of its intralesional and interlesional heterogeneity.

Comparative studies have shown that DCIS has a range of molecular subtypes similar to those identified through gene expression profiling of invasive ductal carcinomas (IDC; refs. 14, 7). Important molecular distinctions are observed in studies of DCIS versus IDC that may provide clues to the origins of molecular diversity in breast carcinogenesis. For example, Allred et al. initially observed that human epidermal growth factor receptor 2 (HER2) is overexpressed at a higher frequency in DCIS than in IDC (15). An increased prevalence of HER2 protein overexpression (and gene amplification) in DCIS versus IDC has been confirmed in a number of subsequent studies (1, 1618). HER2 overexpression seems to be most significant in high-grade DCIS, in which its prevalence is nearly 75% (17). In contrast to this relatively higher prevalence of HER2 overexpression in DCIS, the prevalence of marker expression indicative of the basal-like subtype is relatively lower in DCIS compared with IDC (1, 3, 4, 17, 18).

The apparent imbalance between DCIS and IDC in molecular subtype prevalences suggests that there is no simple linear relationship of progression from DCIS to invasive disease. These observations further suggest that different DCIS subtypes might be associated with different malignant potentials. For example, DCIS with basal marker expression may give rise to invasive carcinoma at a higher frequency than does DCIS exhibiting HER2 overexpression. This lack of a linear relationship might explain the relatively higher frequency of the basal-like subtype recently observed in studies of IDC versus DCIS (2). Alternatively, immunohistochemical detection of protein markers may not always reflect microarray gene expression profiles and therefore may not provide a faithful representation of subtype distribution (7, 19). Most important, immunohistochemistry and microarray studies to define molecular subtypes of DCIS (and even IDC) typically fail to address intralesional heterogeneity, or the possibility of more than one subtype within a single lesion. As discussed below, a more thorough analysis or identification of intralesional heterogeneity may provide new insights into the potential for progression to malignancy.

Examination of individual premalignant lesions illustrates a second level of premalignant heterogeneity, intralesional heterogeneity. The sectored areas of a single lesion can evidence characteristic staining for different subtypes of breast neoplasia. For example, a single lesion might have areas that express proteins associated with a luminal subtype distinct from other areas that express proteins associated with a basal-like subtype (Fig. 1). In addition to cellular manifestations of heterogeneity, intralesional heterogeneity also occurs in nuclear morphologic assessments of nuclear grade, which is an important factor in the clinical assessment of tumors. Components of nuclear morphology that underlie variations in nuclear grade are complex and extend beyond size (20, 21). Variations occur in nuclear shape, contour of nuclear membranes, and number and size of nucleoli. Chromatin can be dispersed or marginated and could form coarse and variably asymmetrical aggregates of heterochromatin. To contend with intralesional heterogeneity at the morphologic level, histologic grading systems assign a score based on properties of selected cell subpopulations. For example, the nuclear grade of a breast IDC is clinically assessed based on nuclear size and shape in areas of highest grade representing as little as 10% of the cell population. Nuclear morphometry, a quantitative approach for assessing heterogeneity of size and other nuclear characteristics, has prognostic value in IDC and DCIS and correlates with nuclear grade (22, 23).

Fig. 1.

Theoretical spectrum of molecular heterogeneity in DCIS. The two ducts represent theoretical comparisons of DCIS between different individuals (i.e., interlesional heterogeneity) or between two regions within a patient's single DCIS lesion (i.e., intralesional heterogeneity). DCIS could range from homogeneity (left) to a surprising degree of heterogeneity (right) within a single lesion. The two ducts depict the ends of a continuum of heterogeneity in nuclear and cell size, presence and number of coexisting molecular subtypes, and genetic and epigenetic alterations.

Fig. 1.

Theoretical spectrum of molecular heterogeneity in DCIS. The two ducts represent theoretical comparisons of DCIS between different individuals (i.e., interlesional heterogeneity) or between two regions within a patient's single DCIS lesion (i.e., intralesional heterogeneity). DCIS could range from homogeneity (left) to a surprising degree of heterogeneity (right) within a single lesion. The two ducts depict the ends of a continuum of heterogeneity in nuclear and cell size, presence and number of coexisting molecular subtypes, and genetic and epigenetic alterations.

Close modal

In an early study of intralesional heterogeneity at the molecular level, Ma et al. observed distinct patterns of gene expression within premalignant and malignant breast lesions that suggested intralesional heterogeneity of histologic grade (24); for example, most grade 2 lesions exhibited a hybrid of grade 1 and grade 3 molecular signatures. More recently, Allred et al. elegantly addressed intralesional diversity at the molecular level in a large series of 120 cases of pure DCIS (25). Almost half of these cases showed variable regions of nuclear grade within an individual biopsy. Intralesional areas of variable histologic grade were microdissected to allow for thorough molecular characterization with a panel of immunohistochemical markers (hormone receptors, GATA3, HER2, p53, and basal cytokeratins) and with gene expression profiling in a subset of samples. A strikingly high proportion (nearly three-quarters) of the DCIS samples with intralesional heterogeneity in nuclear grade showed more than one molecular subtype, with nearly all possible combinations represented (e.g., basal-like and luminal-A subtype in one lesion).

The presence of molecularly and morphologically distinct subpopulations of cells within DCIS raises important questions about the origins of molecular variability and the pathways of breast cancer progression (26). Although DCIS is considered a nonobligate precursor to invasive breast cancer, mathematical models of progression suggest that DCIS and IDC may develop through separate but parallel pathways from a common progenitor (27). Different models that have accurately incorporated progression data collectively suggest multiple pathways of progression (28). Genome-wide profiling of invasive and premalignant lesions of the breast has revealed that the pathway of progression for phenotypically low nuclear grade lesions is molecularly distinct from the pathway for high nuclear grade lesions (10, 29). Low nuclear grade premalignant lesions of the breast include low-grade DCIS, atypical ductal hyperplasia, flat epithelial atypia, and some lobular neoplasias. These lesions all tend to share characteristic alterations such as loss of 16q and gains of chromosome 1q (reviewed in ref. 30). In contrast, high nuclear grade lesions are genetically more complex with varied chromosomal losses and gains. The coexistence of low-grade and high-grade cells in some DCIS lesions therefore suggests the presence of clonal heterogeneity. Mathematical models of progression have not yet considered the implications of intralesional heterogeneity because the primary data typically are derived from established histologic grading systems. Accurate models of progression may depend on consideration of the presence and evolution of clonal diversity. Given that clonal heterogeneity in premalignancy might be an independent risk factor for invasive tumor development (reviewed in ref. 31), future genomic characterization of areas of intralesional heterogeneity within DCIS might also be clinically significant.

To understand the behavioral consequences of intralesional heterogeneity, it is useful to examine the molecular programs that dictate the different phenotypes of DCIS. DCIS lesions are often classified as low, intermediate, or high grade. Several grading systems have been developed and are based on evaluating several phenotypes within the premalignant lesion. The mitotic rate is routinely evaluated in invasive carcinoma and has also been well studied in DCIS. The most commonly evaluated clinical phenotypic features of DCIS are (a) the degree of differentiation, (b) degree of nuclear atypia, and (c) extent of necrosis. The role of the pRb pathway is particularly interesting in the context of histologic grading because the p16/pRb signaling pathway controls a coordinated ensemble of phenotypes that influence grade (i.e., genes that regulate cell cycle progression, differentiation, chromatin remodeling, and cell death). Although not currently used for clinical assessment of DCIS, the status of stress responses in premalignant lesions may provide clinically useful information. These processes contribute to the clinical manifestations of carcinogenesis and may contribute molecular markers that have clinical utility.

The pRb pathway plays a key role in mitotic rate (proliferation), one of the critical phenotypes in determining histologic grade in invasive breast carcinomas. Repression of E2F-dependent transcriptional targets that control cell cycle arrest is assumed to be the major mechanism by which pRb exerts antitumorigenic effects. This focus on cell cycle effects and antiproliferative functions was substantiated by mutation studies showing that removal of pRb allowed accelerated entry into the G1-S phases of the cell cycle and that exogenously introduced unphosphorylated pRb caused cell cycle arrest (3235). This alteration in cell cycle control would be expected to manifest itself via an elevated mitotic rate, such as a higher fraction of cells expressing Ki67, and consequently, via hyperplastic morphologic alterations.

Abrogation of cyclin-dependent kinase 4/6 (CDK4/6), cyclin D1, p16, and/or pRb seems to be required for malignancy, and tumor types are often distinguished by particular alterations in one member of the pathway. Although the abrogation of each member of this pathway imparts tumorigenic potential, emerging evidence in breast carcinogenesis suggests that different mechanisms of p16/pRb pathway inactivation might lead to diverse tumor subtypes (Fig. 2A). For example, cyclin D1 is overexpressed (primarily through gene amplification) predominantly within tumors of the luminal B subtype (2, 3639), whereas basal-like tumors typically show overexpression of p16 and elevated levels of E2F target genes, accompanied by loss of pRb expression (2, 40). Although we do not know if deregulation of pRb signaling (Fig. 2B) has implications for the origins of the basal-like subtype, these data do support that deregulation of pRb is mechanistically linked to it.

Fig. 2.

Distinct molecular subtypes of invasive breast tumors; p16/pRb signaling in breast neoplasia. A, hierarchical clustering (by gene expression) of 130 primary invasive breast tumors identifies distinct molecular subtypes, which are color-coded (top of diagram) as follows: light blue, luminal B; dark blue, luminal A; green, normal-like; orange, basal-like; pink, HER2-positive; gray, unclassified. Loss of functional pRb signaling defines the basal-like subtype of invasive tumors. Expression levels of cyclin D1, p27, pRb, phosphatase and tensin homologue (PTEN), cyclin E, p16, E2F3, Dp2, and AKT3 are shown. The level of expression of each gene is relative to the median expression of this gene across all 130 samples. Adapted from Gauthier et al. (ref. 2; reprinted with permission from Elsevier). B, regulation of p16/pRb signaling is represented diagrammatically. pRb plays an integral role in cell cycle regulation and coordinates multiple signal transduction pathways that regulate proliferation. Deregulation of pRb could occur through multiple mechanisms including loss of heterozygosity, promoter hypermethylation, and gene mutation. Methods of inactivation of the p16/pRb signaling pathway include aberrant phosphorylation through amplification of cyclin E, loss of p27, activation of AKT signaling, and/or loss of PTEN through gene mutation or promoter hypermethylation.

Fig. 2.

Distinct molecular subtypes of invasive breast tumors; p16/pRb signaling in breast neoplasia. A, hierarchical clustering (by gene expression) of 130 primary invasive breast tumors identifies distinct molecular subtypes, which are color-coded (top of diagram) as follows: light blue, luminal B; dark blue, luminal A; green, normal-like; orange, basal-like; pink, HER2-positive; gray, unclassified. Loss of functional pRb signaling defines the basal-like subtype of invasive tumors. Expression levels of cyclin D1, p27, pRb, phosphatase and tensin homologue (PTEN), cyclin E, p16, E2F3, Dp2, and AKT3 are shown. The level of expression of each gene is relative to the median expression of this gene across all 130 samples. Adapted from Gauthier et al. (ref. 2; reprinted with permission from Elsevier). B, regulation of p16/pRb signaling is represented diagrammatically. pRb plays an integral role in cell cycle regulation and coordinates multiple signal transduction pathways that regulate proliferation. Deregulation of pRb could occur through multiple mechanisms including loss of heterozygosity, promoter hypermethylation, and gene mutation. Methods of inactivation of the p16/pRb signaling pathway include aberrant phosphorylation through amplification of cyclin E, loss of p27, activation of AKT signaling, and/or loss of PTEN through gene mutation or promoter hypermethylation.

Close modal

Although undoubtedly important to neoplasia, the mitotic rate is not the only phenotype providing prognostic information that is influenced by abrogation of the pRb pathway. For example, it is becoming widely appreciated that the pRb protein integrates cell cycle arrest with cellular differentiation. Both in vitro and in vivo model systems have provided evidence that pRb plays a role in determining cell fate (4143). Work in a growing number of tissues has shown that functional expression of pRb transcriptionally modulates genes that are important in achieving complete cellular differentiation. These regulatory properties seem to be independent of pRb effects on cell cycle progression. For example, pRb mutants that uncouple cell cycle properties from differentiation effects have been identified (44).

In normal cells, the pRb protein is upregulated during differentiation and interacts with selected coregulators of transcription to activate or repress gene loci. Absence of pRb function allows morphologic differentiation but prevents full functional maturation of cells. In human breast epithelial cells, removal of pRb function allows cells to form characteristic acinar structures in three-dimensional culture conditions, but the expression of lactoferrin and serum cytokeratin 19 (CK19), which are markers for luminal differentiation, are not expressed (45). Molecular studies have shown that the upregulation of pRb transcription in some tissue systems is regulated by p53 and by mechanical forces in the surrounding stroma. Manipulation of these conditions could induce lactoferrin production in human mammary epithelial cells (46). The consequences of these regulatory circuits in breast premalignancy are interesting. In tumors in which the pRb pathway has been abrogated by mutation of other members of the pathway, e.g., CDK4 mutation, p16 silencing or cyclin D1 overexpression, inactivation of pRb interactions with E2F would still affect cell cycle arrest and allow an increased mitotic rate. However, tumors with these mutational alterations would still express pRb protein, which could coregulate genes important in differentiation. In contrast, basal-like tumors, which are missing pRb function, not only experience a loss of cell cycle control but also fail to express markers of cellular differentiation.

Recent studies indicate that pRb may also play a pivotal role in the generation of nuclear atypia through the regulation of genetic and epigenetic alterations and changes in cellular polarity. Nuclei of an abnormal shape, size, and texture are a hallmark of malignancy. These nuclear properties are governed by the packaging of chromatin within the nucleus and the proper partitioning of chromosomal elements. Epigenetic systems that dictate chromatin structure are regulated by pRb, most often through the downstream regulation of transcriptional targets that activate histone methyltransferases, DNA methyltransferases, or acetylases. The repression of p16 activity, accompanied by the inactivation of pRb as a transcriptional repressor, leads to the overexpression of chromatin-remodeling proteins such as EZH2 and SUZ12 (47, 48). In addition to executing the silencing of selected loci, overexpression of these proteins has been identified in approximately half of DCIS lesions and indicate a poor prognosis when detected in invasive breast cancers (49, 50). In addition to epigenetic controls on nuclear size and shape, genetic changes may also play an important role in generating nuclear atypia. The role of the pRb pathway in disrupting genomic integrity is discussed below. Last, coordination of centrosome duplication with DNA synthesis is choreographed by the pRb pathway. Abrogation of this pathway leads to the generation of aneuploidy and the loss of proper polarity (51).

Potential mechanisms of pRb pathway alterations, including deletion, mutation, and/or promoter hypermethylation, may have differential effects on the nucleus. Studies by our group and others show that pRb gene expression levels are lowest in basal-like tumors (2, 40, 52, 53). Tumors with loss of functional pRb signaling have a higher frequency of chromosome copy number alterations than do tumors with aberrations of other members of the p16/pRb pathway (54).

Finally, in the assessment of DCIS lesions, increased cell death is associated with a more aggressive behavior. The control of cell death pathways is complex, but studies have linked pRb alterations with the control of p53 function and the regulation of cell death (55). Therefore, alteration of the pRb pathway modulates multiple phenotypes, including proliferation indices, differentiation, nuclear atypia, and death indices that are important in malignancy. A careful analysis of the mechanisms of deregulating this pathway may provide useful clinical information.

In addition to the typical phenotypes used to classify DCIS in the clinic, several molecular markers might prove useful in stratifying risk for future invasive tumor formation and may also relate to pRb biology. There is a notable link between the pRb pathway and the activation of cellular senescence. Although well-studied in vitro, senescent cells are just beginning to be studied in vivo, in which the understanding of their presence and significance is rapidly emerging. Studies in human samples (2) and murine models of prostate carcinogenesis (56), melanocytic nevi (5759), lung adenomas (60, 61), and lymphoma (62) show that overexpression of p16 protein and other markers of cellular senescence preferentially identify indolent premalignant and benign lesions. In studying p16/pRb signaling within premalignant lesions such as DCIS and atypical ductal hyperplasia, it is important to consider that p16 overexpression is conditional on the functional state of pRb. Unlike cells of most malignant tumors, cells within premalignant lesions may have intact pRb signaling. The p16 overexpression in premalignancy may be a physiologic response to stress activation leading to cell cycle arrest and a senescence program. Indications of an intact response to cellular stress in DCIS seems to be of clinical significance because overexpression of p16 in the absence of proliferation (i.e., activation of a senescence program) identifies women less likely to develop subsequent breast cancer (2). These observations support the hypothesis that cellular senescence is a barrier to tumorigenesis, which is maintained in some premalignant lesions.

In normal cells, activation of the pRb pathway in response to cellular aging or cellular stress such as oxidative, metabolic, hypoxic, and/or oncogenic stress drives cell cycle arrest and plays an important role in premature and replicative senescence-response pathways (63, 64). Through inhibition of CDK4/6, p16 blocks the phosphorylation of pRb and thus inhibits E2F transcription factors from binding to promoter sequences essential for cell cycle progression (6569). Furthermore, the pRb pathway regulates the expression of stress proteins known to be important in conferring malignant phenotypes. Human mammary epithelial cells typically do not express the proinflammatory protein cyclooxygenase-2 (COX-2). However, mutations within the pRb pathway affect the expression of this protein in distinct ways. As recently reported, abrogation of the pRb pathway through p16 repression enhanced the induction of COX-2 when cells were exposed to a variety of exogenous stresses (2). In contrast, cyclin D1 overexpression did not sensitize cells to the stress signals and did not induce COX-2. Of interest, removal of pRb function itself caused a constitutive overexpression of COX-2. It is intriguing to consider that subpopulations of normal human mammary epithelial cells in vivo and in vitro show p16/pRb inactivation through promoter hypermethylation, and consequently, COX-2 overexpression (70, 71). Because COX-2 enhances cell migration, angiogenesis, and proliferation and inhibits apoptosis and immune surveillance, it is clear that different mutational mechanisms for abrogating the pRb pathway have important phenotypic consequences for tumors.

Premalignant cells that have abrogated pRb signaling and bypass stress-induced senescence may also manifest overexpression of p16. In this setting, elevated levels of p16 protein in highly proliferative cells suggest that the growth-suppressive effects of p16 are short-circuited downstream of p16. Despite exhibiting hallmarks of stress activation, such as overexpression of p16, p53, p21, and COX-2 and phosphorylation activation of stress-activated kinases such as p38, which are usually associated with the induction of a proliferative arrest, cells with dysfunctional pRb undergo unobstructed proliferation (2, 55, 72). An abrogated response to stress signals in DCIS also seems to have clinical value because overexpression of p16 in the presence of ongoing proliferation identifies women more likely to develop subsequent breast cancer (Fig. 3).

Fig. 3.

p16 Overexpression coupled with proliferation increases the risk of subsequent tumor events among women with DCIS. A, immunohistochemistry staining for p16 and Ki67 in normal breast tissue (n = 40), atypical ductal hyperplasia (ADH; n = 33), low-, intermediate (Int)-, and high-grade DCIS (n = 142), and IDC (n = 100). Each bar graph represents the percentage of immunopositive cases. p16 staining was scored on a 0, 1, 2, 3 scale based on the extent of immunopositive cells (0, no staining; 1, <25%; 2, 25-75%; 3, >75%). Positive/high p16 staining is defined as a score of ≥2. The Ki67 index was determined by manually counting a minimum of 1,000 nuclei within at least three 40× fields. Positive/high Ki67 is defined as greater than 10% immunopositive cells. B, Kaplan-Meier plots of recurrence-free survival by p16 status, Ki67 status, and p16/Ki67 coexpression status among 70 patients diagnosed with and treated for pure DCIS. Adapted from Gauthier et al. (2); reprinted with permission from Elsevier.

Fig. 3.

p16 Overexpression coupled with proliferation increases the risk of subsequent tumor events among women with DCIS. A, immunohistochemistry staining for p16 and Ki67 in normal breast tissue (n = 40), atypical ductal hyperplasia (ADH; n = 33), low-, intermediate (Int)-, and high-grade DCIS (n = 142), and IDC (n = 100). Each bar graph represents the percentage of immunopositive cases. p16 staining was scored on a 0, 1, 2, 3 scale based on the extent of immunopositive cells (0, no staining; 1, <25%; 2, 25-75%; 3, >75%). Positive/high p16 staining is defined as a score of ≥2. The Ki67 index was determined by manually counting a minimum of 1,000 nuclei within at least three 40× fields. Positive/high Ki67 is defined as greater than 10% immunopositive cells. B, Kaplan-Meier plots of recurrence-free survival by p16 status, Ki67 status, and p16/Ki67 coexpression status among 70 patients diagnosed with and treated for pure DCIS. Adapted from Gauthier et al. (2); reprinted with permission from Elsevier.

Close modal

In support of pRb biology linking multiple malignant phenotypes, tumors with the lowest expression of pRb exhibit illuminating characteristics. Increased immunodetection of p16 correlates with decreased pRb levels in breast carcinomas (2, 53, 73, 74). Tumors with p16 overexpression at the immunohistochemical level have been identified as poorly differentiated tumors through associations with high nuclear grade, high Ki67, increased p53, and low estrogen receptor/progesterone receptor expression (7579). Of importance, these tumors also overexpress COX-2. Therefore, although the activation or inactivation of cellular stress and senescence pathways are not currently used clinically, this information may be helpful in the future.

The acquisition of genomic instability through the deregulation of p16/pRb signaling provides the potential for generating heterogeneity. Despite the well-established role of pRb dysfunction in deregulating cell cycle progression and desensitizing cells to damage-induced cell cycle arrest or apoptotic signals, only more recent studies have directly addressed the causal relationship between loss of pRb signaling and genomic instability. The loss of pRb deregulates the expression of genes involved in mitotic progression and chromosome segregation generating genetic diversity. Furthermore, the loss of functional pRb not only leads to inappropriate proliferation but also to whole-chromosome gains and losses (aneuploidy) due to aberrant expression of spindle checkpoint proteins such as mitotic arrest-deficient 2 (MAD2; refs. 80, 81). It is interesting to note that loss of p16 expression also results in chromosome instability and aneuploidy but through a pRb-independent mechanism. Indeed, it was recently shown that loss of p16 expression in genomically stable, normal mammary epithelial cells results in the production of supernumerary centrosomes in a CDK2/p21-dependent fashion (51). These collective results suggest that the loss of p16/pRb signaling may cause chromosome and genomic instability early during tumorigenesis (i.e., prior to the acquisition of oncogenic events) and might contribute to the genesis of molecular heterogeneity in tumors.

However, differences in marker expression might represent the end result of increased mutations or, alternatively, may represent the emerging result of ecological interactions. Consistent with this speculation, our studies of DCIS (2) found that heterogeneity was the prevalent phenotype, with few lesions showing homogeneous expression of a given marker. In DCIS lesions expressing a senescence signature, differences in the extent of p16 expression might represent regional differences in cellular stress or different “snapshots in time” of the stress response rather than the consequence of mutation.

Last, concepts ordinarily used in evolutionary or ecological studies might provide insights into the functional significance of heterogeneity in premalignant lesions. The significance of intralesional heterogeneity may be analogous to that of the generation of diversity observed in ecological interactions developed by stressed populations. In these populations, exposure to stress activates evolutionary and ecological interactions that result in diversity which buffers against complete loss of viability under rapidly changing conditions (i.e., the insurance hypothesis; ref. 82). Conceptually speaking, the generation of heterogeneity therefore serves a function that might extend beyond direct induction of mutational changes, serving, for example, to preserve the survival of the population as a whole.

No potential conflicts of interest were disclosed.

We thank Dr. Philippe Gascard for helpful editing of this manuscript.

1
Livasy
CA
,
Perou
CM
,
Karaca
G
, et al
. 
Identification of a basal-like subtype of breast ductal carcinoma in situ
.
Hum Pathol
2007
;
38
:
197
204
.
2
Gauthier
ML
,
Berman
HK
,
Miller
C
, et al
. 
Abrogated response to cellular stress identifies DCIS associated with subsequent tumor events and defines basal-like breast tumors
.
Cancer Cell
2007
;
12
:
479
91
.
3
Dabbs
DJ
,
Chivukula
M
,
Carter
G
,
Bhargava
R
. 
Basal phenotype of ductal carcinoma in situ: recognition and immunohistologic profile
.
Mod Pathol
2006
;
19
:
1506
11
.
4
Bryan
BB
,
Schnitt
SJ
,
Collins
LC
. 
Ductal carcinoma in situ with basal-like phenotype: a possible precursor to invasive basal-like breast cancer
.
Mod Pathol
2006
;
19
:
617
21
.
5
Ellsworth
RE
,
Vertrees
A
,
Love
B
,
Hooke
JA
,
Ellsworth
DL
,
Shriver
CD
. 
Chromosomal alterations associated with the transition from in situ to invasive breast cancer
.
Ann Surg Oncol
2008
;
15
:
2519
25
.
6
Iakovlev
VV
,
Arneson
NC
,
Wong
V
, et al
. 
Genomic differences between pure ductal carcinoma in situ of the breast and that associated with invasive disease: a calibrated aCGH study
.
Clin Cancer Res
2008
;
14
:
4446
54
.
7
Hannemann
J
,
Velds
A
,
Halfwerk
JB
,
Kreike
B
,
Peterse
JL
,
van de Vijver
MJ
. 
Classification of ductal carcinoma in situ by gene expression profiling
.
Breast Cancer Res
2006
;
8
:
R61
.
8
Schuetz
CS
,
Bonin
M
,
Clare
SE
, et al
. 
Progression-specific genes identified by expression profiling of matched ductal carcinomas in situ and invasive breast tumors, combining laser capture microdissection and oligonucleotide microarray analysis
.
Cancer Res
2006
;
66
:
5278
86
.
9
Hwang
ES
,
DeVries
S
,
Chew
KL
, et al
. 
Patterns of chromosomal alterations in breast ductal carcinoma in situ
.
Clin Cancer Res
2004
;
10
:
5160
7
.
10
Buerger
H
,
Otterbach
F
,
Simon
R
, et al
. 
Comparative genomic hybridization of ductal carcinoma in situ of the breast—evidence of multiple genetic pathways
.
J Pathol
1999
;
187
:
396
402
.
11
Finak
G
,
Bertos
N
,
Pepin
F
, et al
. 
Stromal gene expression predicts clinical outcome in breast cancer
.
Nat Med
2008
;
14
:
518
27
.
12
Ma
XJ
,
Dahiya
S
,
Richardson
E
,
Erlander
M
,
Sgroi
DC
. 
Gene expression profiling of the tumor microenvironment during breast cancer progression
.
Breast Cancer Res
2009
;
11
:
R7
.
13
Trimboli
AJ
,
Cantemir-Stone
CZ
,
Li
F
, et al
. 
Pten in stromal fibroblasts suppresses mammary epithelial tumours
.
Nature
2009
;
461
:
1084
91
.
14
Polyak
K
,
Haviv
I
,
Campbell
IG
. 
Co-evolution of tumor cells and their microenvironment
.
Trends Genet
2009
;
25
:
30
8
.
15
Allred
DC
,
Clark
GM
,
Molina
R
, et al
. 
Overexpression of HER-2/neu and its relationship with other prognostic factors change during the progression of in situ to invasive breast cancer
.
Hum Pathol
1992
;
23
:
974
9
.
16
Latta
EK
,
Tjan
S
,
Parkes
RK
,
O'Malley
FP
. 
The role of HER2/neu overexpression/amplification in the progression of ductal carcinoma in situ to invasive carcinoma of the breast
.
Mod Pathol
2002
;
15
:
1318
25
.
17
Meijnen
P
,
Peterse
JL
,
Antonini
N
,
Rutgers
EJ
,
van de Vijver
MJ
. 
Immunohistochemical categorisation of ductal carcinoma in situ of the breast
.
Br J Cancer
2008
;
98
:
137
42
.
18
Tamimi
RM
,
Baer
HJ
,
Marotti
J
, et al
. 
Comparison of molecular phenotypes of ductal carcinoma in situ and invasive breast cancer
.
Breast Cancer Res
2008
;
10
:
R67
.
19
Nielsen
TO
,
Hsu
FD
,
Jensen
K
, et al
. 
Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma
.
Clin Cancer Res
2004
;
10
:
5367
74
.
20
Black
MM
,
Speer
FD
. 
Nuclear structure in mammary carcinoma
.
Am J Pathol
1950
;
26
:
913
21
.
21
Zink
D
,
Fischer
AH
,
Nickerson
JA
. 
Nuclear structure in cancer cells
.
Nat Rev Cancer
2004
;
4
:
677
87
.
22
Baak
JP
,
Van Dop
H
,
Kurver
PH
,
Hermans
J
. 
The value of morphometry to classic prognosticators in breast cancer
.
Cancer
1985
;
56
:
374
82
.
23
Tan
PH
,
Goh
BB
,
Chiang
G
,
Bay
BH
. 
Correlation of nuclear morphometry with pathologic parameters in ductal carcinoma in situ of the breast
.
Mod Pathol
2001
;
14
:
937
41
.
24
Ma
XJ
,
Salunga
R
,
Tuggle
JT
, et al
. 
Gene expression profiles of human breast cancer progression
.
Proc Natl Acad Sci U S A
2003
;
100
:
5974
9
.
25
Allred
DC
,
Wu
Y
,
Mao
S
, et al
. 
Ductal carcinoma in situ and the emergence of diversity during breast cancer evolution
.
Clin Cancer Res
2008
;
14
:
370
8
.
26
Polyak
K
. 
Breast cancer: origins and evolution
.
J Clin Invest
2007
;
117
:
3155
63
.
27
Sontag
L
,
Axelrod
DE
. 
Evaluation of pathways for progression of heterogeneous breast tumors
.
J Theor Biol
2005
;
232
:
179
89
.
28
Lin
S
. 
Mixture modeling of progression pathways of heterogeneous breast tumors
.
J Theor Biol
2007
;
249
:
254
61
.
29
Simpson
PT
,
Reis-Filho
JS
,
Gale
T
,
Lakhani
SR
. 
Molecular evolution of breast cancer
.
J Pathol
2005
;
205
:
248
54
.
30
Abdel-Fatah
TM
,
Powe
DG
,
Hodi
Z
,
Lee
AH
,
Reis-Filho
JS
,
Ellis
IO
. 
High frequency of coexistence of columnar cell lesions, lobular neoplasia, and low grade ductal carcinoma in situ with invasive tubular carcinoma and invasive lobular carcinoma
.
Am J Surg Pathol
2007
;
31
:
417
26
.
31
Merlo
LM
,
Pepper
JW
,
Reid
BJ
,
Maley
CC
. 
Cancer as an evolutionary and ecological process
.
Nat Rev Cancer
2006
;
6
:
924
35
.
32
Knudsen
ES
,
Wang
JY
. 
Differential regulation of retinoblastoma protein function by specific Cdk phosphorylation sites
.
J Biol Chem
1996
;
271
:
8313
20
.
33
Herrera
RE
,
Sah
VP
,
Williams
BO
,
Makela
TP
,
Weinberg
RA
,
Jacks
T
. 
Altered cell cycle kinetics, gene expression, and G1 restriction point regulation in Rb-deficient fibroblasts
.
Mol Cell Biol
1996
;
16
:
2402
7
.
34
Lundberg
AS
,
Weinberg
RA
. 
Functional inactivation of the retinoblastoma protein requires sequential modification by at least two distinct cyclin-cdk complexes
.
Mol Cell Biol
1998
;
18
:
753
61
.
35
White
RJ
,
Trouche
D
,
Martin
K
,
Jackson
SP
,
Kouzarides
T
. 
Repression of RNA polymerase III transcription by the retinoblastoma protein
.
Nature
1996
;
382
:
88
90
.
36
Agarwal
R
,
Gonzalez-Angulo
AM
,
Myhre
S
, et al
. 
Integrative analysis of cyclin protein levels identifies cyclin b1 as a classifier and predictor of outcomes in breast cancer
.
Clin Cancer Res
2009
;
15
:
3654
62
.
37
Aaltonen
K
,
Amini
RM
,
Landberg
G
, et al
. 
Cyclin D1 expression is associated with poor prognostic features in estrogen receptor positive breast cancer
.
Breast Cancer Res Treat
2009
;
113
:
75
82
.
38
Elsheikh
S
,
Green
AR
,
Aleskandarany
MA
, et al
. 
CCND1 amplification and cyclin D1 expression in breast cancer and their relation with proteomic subgroups and patient outcome
.
Breast Cancer Res Treat
2008
;
109
:
325
35
.
39
Loden
M
,
Stighall
M
,
Nielsen
NH
, et al
. 
The cyclin D1 high and cyclin E high subgroups of breast cancer: separate pathways in tumorogenesis based on pattern of genetic aberrations and inactivation of the pRb node
.
Oncogene
2002
;
21
:
4680
90
.
40
Herschkowitz
JI
,
He
X
,
Fan
C
,
Perou
CM
. 
The functional loss of the retinoblastoma tumor suppressor is a common event in Basal-like and Luminal B breast carcinomas
.
Breast Cancer Res
2008
;
10
:
R75
.
41
Classon
M
,
Harlow
E
. 
The retinoblastoma tumour suppressor in development and cancer
.
Nat Rev Cancer
2002
;
2
:
910
7
.
42
Khidr
L
,
Chen
PL
. 
RB, the conductor that orchestrates life, death and differentiation
.
Oncogene
2006
;
25
:
5210
9
.
43
Burkhart
DL
,
Sage
J
. 
Cellular mechanisms of tumour suppression by the retinoblastoma gene
.
Nat Rev Cancer
2008
;
8
:
671
82
.
44
Sellers
WR
,
Novitch
BG
,
Miyake
S
, et al
. 
Stable binding to E2F is not required for the retinoblastoma protein to activate transcription, promote differentiation, and suppress tumor cell growth
.
Genes Dev
1998
;
12
:
95
106
.
45
Spancake
KM
,
Anderson
CB
,
Weaver
VM
,
Matsunami
N
,
Bissell
MJ
,
White
RL
. 
E7-transduced human breast epithelial cells show partial differentiation in three-dimensional culture
.
Cancer Res
1999
;
59
:
6042
5
.
46
Close
MJ
,
Howlett
AR
,
Roskelley
CD
, et al
. 
Lactoferrin expression in mammary epithelial cells is mediated by changes in cell shape and actin cytoskeleton
.
J Cell Sci
1997
;
110
:
2861
71
.
47
Reynolds
PA
,
Sigaroudinia
M
,
Zardo
G
, et al
. 
Tumor suppressor p16INK4A regulates polycomb-mediated DNA hypermethylation in human mammary epithelial cells
.
J Biol Chem
2006
;
281
:
24790
802
.
48
Bracken
AP
,
Pasini
D
,
Capra
M
,
Prosperini
E
,
Colli
E
,
Helin
K
. 
EZH2 is downstream of the pRB-E2F pathway, essential for proliferation and amplified in cancer
.
EMBO J
2003
;
22
:
5323
35
.
49
Ding
L
,
Erdmann
C
,
Chinnaiyan
AM
,
Merajver
SD
,
Kleer
CG
. 
Identification of EZH2 as a molecular marker for a precancerous state in morphologically normal breast tissues
.
Cancer Res
2006
;
66
:
4095
9
.
50
Kleer
CG
,
Cao
Q
,
Varambally
S
, et al
. 
EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells
.
Proc Natl Acad Sci U S A
2003
;
100
:
11606
11
.
51
McDermott
KM
,
Zhang
J
,
Holst
CR
,
Kozakiewicz
BK
,
Singla
V
,
Tlsty
TD
. 
p16(INK4a) prevents centrosome dysfunction and genomic instability in primary cells
.
PLoS Biol
2006
;
4
:
e51
.
52
Bieche
I
,
Lidereau
R
. 
Loss of heterozygosity at 13q14 correlates with RB1 gene underexpression in human breast cancer
.
Mol Carcinog
2000
;
29
:
151
8
.
53
Nielsen
NH
,
Emdin
SO
,
Cajander
J
,
Landberg
G
. 
Deregulation of cyclin E and D1 in breast cancer is associated with inactivation of the retinoblastoma protein
.
Oncogene
1997
;
14
:
295
304
.
54
Fridlyand
J
,
Snijders
AM
,
Ylstra
B
, et al
. 
Breast tumor copy number aberration phenotypes and genomic instability
.
BMC Cancer
2006
;
6
:
96
.
55
Zhang
J
,
Pickering
CR
,
Holst
CR
,
Gauthier
ML
,
Tlsty
TD
. 
p16INK4a modulates p53 in primary human mammary epithelial cells
.
Cancer Res
2006
;
66
:
10325
31
.
56
Chen
Z
,
Trotman
LC
,
Shaffer
D
, et al
. 
Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis
.
Nature
2005
;
436
:
725
30
.
57
Michaloglou
C
,
Vredeveld
LC
,
Soengas
MS
, et al
. 
BRAFE600-associated senescence-like cell cycle arrest of human naevi
.
Nature
2005
;
436
:
720
4
.
58
Gray-Schopfer
VC
,
Cheong
SC
,
Chong
H
, et al
. 
Cellular senescence in naevi and immortalisation in melanoma: a role for p16?
Br J Cancer
2006
;
95
:
496
505
.
59
Wajapeyee
N
,
Serra
RW
,
Zhu
X
,
Mahalingam
M
,
Green
MR
. 
Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7
.
Cell
2008
;
132
:
363
74
.
60
Dankort
D
,
Filenova
E
,
Collado
M
,
Serrano
M
,
Jones
K
,
McMahon
M
. 
A new mouse model to explore the initiation, progression, and therapy of BRAFV600E-induced lung tumors
.
Genes Dev
2007
;
21
:
379
84
.
61
Collado
M
,
Gil
J
,
Efeyan
A
, et al
. 
Tumour biology: senescence in premalignant tumours
.
Nature
2005
;
436
:
642
.
62
Braig
M
,
Lee
S
,
Loddenkemper
C
, et al
. 
Oncogene-induced senescence as an initial barrier in lymphoma development
.
Nature
2005
;
436
:
660
5
.
63
Serrano
M
,
Gomez-Lahoz
E
,
DePinho
RA
,
Beach
D
,
Bar-Sagi
D
. 
Inhibition of ras-induced proliferation and cellular transformation by p16INK4
.
Science
1995
;
267
:
249
52
.
64
Serrano
M
,
Lee
H
,
Chin
L
,
Cordon-Cardo
C
,
Beach
D
,
DePinho
RA
. 
Role of the INK4a locus in tumor suppression and cell mortality
.
Cell
1996
;
85
:
27
37
.
65
Koh
J
,
Enders
GH
,
Dynlacht
BD
,
Harlow
E
. 
Tumour-derived p16 alleles encoding proteins defective in cell-cycle inhibition
.
Nature
1995
;
375
:
506
10
.
66
Dyson
N
. 
The regulation of E2F by pRB-family proteins
.
Genes Dev
1998
;
12
:
2245
62
.
67
Nevins
JR
. 
Toward an understanding of the functional complexity of the E2F and retinoblastoma families
.
Cell Growth Differ
1998
;
9
:
585
93
.
68
Harbour
JW
,
Dean
DC
. 
Rb function in cell-cycle regulation and apoptosis
.
Nat Cell Biol
2000
;
2
:
E65
7
.
69
Lukas
J
,
Parry
D
,
Aagaard
L
, et al
. 
Retinoblastoma-protein-dependent cell-cycle inhibition by the tumour suppressor p16
.
Nature
1995
;
375
:
503
6
.
70
Crawford
YG
,
Gauthier
ML
,
Joubel
A
, et al
. 
Histologically normal human mammary epithelia with silenced p16(INK4a) overexpress COX-2, promoting a premalignant program
.
Cancer Cell
2004
;
5
:
263
73
.
71
Holst
CR
,
Nuovo
GJ
,
Esteller
M
, et al
. 
Methylation of p16(INK4a) promoters occurs in vivo in histologically normal human mammary epithelia
.
Cancer Res
2003
;
63
:
1596
601
.
72
Gauthier
ML
,
Pickering
CR
,
Miller
CJ
, et al
. 
p38 regulates cyclooxygenase-2 in human mammary epithelial cells and is activated in premalignant tissue
.
Cancer Res
2005
;
65
:
1792
9
.
73
Dublin
EA
,
Patel
NK
,
Gillett
CE
,
Smith
P
,
Peters
G
,
Barnes
DM
. 
Retinoblastoma and p16 proteins in mammary carcinoma: their relationship to cyclin D1 and histopathological parameters
.
Int J Cancer
1998
;
79
:
71
5
.
74
Gorgoulis
VG
,
Koutroumbi
EN
,
Kotsinas
A
, et al
. 
Alterations of p16-pRb pathway and chromosome locus 9p21-22 in sporadic invasive breast carcinomas
.
Mol Med
1998
;
4
:
807
22
.
75
Emig
R
,
Magener
A
,
Ehemann
V
, et al
. 
Aberrant cytoplasmic expression of the p16 protein in breast cancer is associated with accelerated tumour proliferation
.
Br J Cancer
1998
;
78
:
1661
8
.
76
Hui
R
,
Macmillan
RD
,
Kenny
FS
, et al
. 
INK4a gene expression and methylation in primary breast cancer: overexpression of p16INK4a messenger RNA is a marker of poor prognosis
.
Clin Cancer Res
2000
;
6
:
2777
87
.
77
Han
S
,
Ahn
SH
,
Park
K
, et al
. 
P16INK4a protein expression is associated with poor survival of the breast cancer patients after CMF chemotherapy
.
Breast Cancer Res Treat
2001
;
70
:
205
12
.
78
Milde-Langosch
K
,
Bamberger
AM
,
Rieck
G
,
Kelp
B
,
Loning
T
. 
Overexpression of the p16 cell cycle inhibitor in breast cancer is associated with a more malignant phenotype
.
Breast Cancer Res Treat
2001
;
67
:
61
70
.
79
Singh
M
,
Parnes
MB
,
Spoelstra
N
,
Bleile
MJ
,
Robinson
WA
. 
p16 expression in sentinel nodes with metastatic breast carcinoma: evaluation of its role in developing triaging strategies for axillary node dissection and a marker of poor prognosis
.
Hum Pathol
2004
;
35
:
1524
30
.
80
Hernando
E
,
Nahle
Z
,
Juan
G
, et al
. 
Rb inactivation promotes genomic instability by uncoupling cell cycle progression from mitotic control
.
Nature
2004
;
430
:
797
802
.
81
Sotillo
R
,
Hernando
E
,
Diaz-Rodriguez
E
, et al
. 
Mad2 overexpression promotes aneuploidy and tumorigenesis in mice
.
Cancer Cell
2007
;
11
:
9
23
.
82
Yachi
S
,
Loreau
M
. 
Biodiversity and ecosystem productivity in a fluctuating environment: the insurance hypothesis
.
Proc Natl Acad Sci U S A
1999
;
96
:
1463
8
.